Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Biochem J ; 474(23): 3985-3999, 2017 11 21.
Article in English | MEDLINE | ID: mdl-29074640

ABSTRACT

The 18 kDa translocator protein (TSPO) is a ubiquitous conserved outer mitochondrial membrane protein implicated in numerous cell and tissue functions, including steroid hormone biosynthesis, respiration, cell proliferation, and apoptosis. TSPO binds with high affinity to cholesterol and numerous compounds, is expressed at high levels in steroid-synthesizing tissues, and mediates cholesterol import into mitochondria, which is the rate-limiting step in steroid formation. In humans, the rs6971 polymorphism on the TSPO gene leads to an amino acid substitution in the fifth transmembrane loop of the protein, which is where the cholesterol-binding domain of TSPO is located, and this polymorphism has been associated with anxiety-related disorders. However, recent knockout mouse models have provided inconsistent conclusions of whether TSPO is directly involved in steroid synthesis. In this report, we show that TSPO deletion mutations in rat and its corresponding rs6971 polymorphism in humans alter adrenocorticotropic hormone-induced plasma corticosteroid concentrations. Rat tissues examined show increased cholesteryl ester accumulation, and neurosteroid formation was undetectable in homozygous rats. These results also support a role for TSPO ligands in diseases with steroid-dependent stress and anxiety elements.


Subject(s)
Adrenocorticotropic Hormone/pharmacology , Carrier Proteins/genetics , Hydrocortisone/blood , Polymorphism, Single Nucleotide , Receptors, GABA-A/genetics , Receptors, GABA/genetics , Adolescent , Adult , Animals , Base Sequence , Carrier Proteins/metabolism , Cholesterol Esters/biosynthesis , Cholesterol Esters/blood , Chorionic Gonadotropin/pharmacology , Cloning, Molecular , Corticosterone/biosynthesis , Corticosterone/blood , Embryo, Mammalian , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Humans , Hydrocortisone/biosynthesis , Male , Plasmids/chemistry , Plasmids/metabolism , Pregnanolone/biosynthesis , Pregnanolone/blood , Rats , Rats, Transgenic , Receptors, GABA/metabolism , Receptors, GABA-A/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Testosterone/biosynthesis , Testosterone/blood , Zinc Fingers , Zygote/drug effects , Zygote/growth & development , Zygote/metabolism
2.
Article in English | MEDLINE | ID: mdl-27524977

ABSTRACT

The adrenal cortex is critical for physiological function as the central site of glucocorticoid and mineralocorticoid synthesis. It possesses a great degree of specialized compartmentalization at multiple hierarchical levels, ranging from the tissue down to the molecular levels. In this paper, we discuss this functionalization, beginning with the tissue zonation of the adrenal cortex and how this impacts steroidogenic output. We then discuss the cellular biology of steroidogenesis, placing special emphasis on the mitochondria. Mitochondria are classically known as the "powerhouses of the cell" for their central role in respiratory adenosine triphosphate synthesis, and attention is given to mitochondrial electron transport, in both the context of mitochondrial respiration and mitochondrial steroid metabolism. Building on work demonstrating functional assembly of large protein complexes in respiration, we further review research demonstrating a role for multimeric protein complexes in mitochondrial cholesterol transport, steroidogenesis, and mitochondria-endoplasmic reticulum contact. We aim to highlight with this review the shift in steroidogenic cell biology from a focus on the actions of individual proteins in isolation to the actions of protein assemblies working together to execute cellular functions.

4.
Biochem Soc Trans ; 43(4): 572-8, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26551695

ABSTRACT

The translocator protein (TSPO; 18k Da) is an evolutionarily conserved outer mitochondrial membrane (OMM) protein highly expressed in steroid-synthesizing cells and found to possess a number of physiological and drug-binding partners. Extensive pharmacological, biochemical and cell biological research over the years has led to a model of TSPO involvement in mitochondrial cholesterol transport and promotion of steroid synthesis, a model guiding the design of drugs useful in stimulating neurosteroid synthesis and alleviating psychopathological symptoms. The involvement of TSPO in these processes has been called into question; however, with the publication of TSPO-deletion mouse models which saw no changes in steroid production. Here, we review work characterizing TSPO in steroidogenesis and offer perspective to research into TSPO pharmacology and its involvement in steroid biosynthesis.


Subject(s)
Cholesterol/metabolism , Receptors, GABA/metabolism , Steroids/biosynthesis , Animals , Benzodiazepines/pharmacology , Binding Sites/drug effects , Humans , Isoquinolines/pharmacology , Ligands , Models, Molecular , Receptors, GABA/chemistry
5.
Proc Natl Acad Sci U S A ; 112(23): 7261-6, 2015 Jun 09.
Article in English | MEDLINE | ID: mdl-26039990

ABSTRACT

Translocator protein (TSPO) is a key member of the mitochondrial cholesterol transport complex in steroidogenic tissues. To assess the function of TSPO, we generated two lines of Cre-mediated Tspo conditional knockout (cKO) mice. First, gonadal somatic cell-targeting Amhr2-Cre mice were crossed with Tspo-floxed mice to obtain F1 Tspo Amhr2 cKO mice (Tspo(fl/fl);Amhr2-Cre(/+)). The unexpected Mendelian ratio of 4.4% cKO mice was confirmed by genotyping of 12.5-day-postcoitum (dpc) embryos. As Amhr2-Cre is expressed in gonads at 12.5 dpc, these findings suggest preimplantation selection of embryos. Analysis of expression databases revealed elevated levels of Amhr2 in two- and eight-cell zygotes, suggesting ectopic Tspo silencing before the morula stage and demonstrating elevated embryonic lethality and involvement of TSPO in embryonic development. To circumvent this issue, steroidogenic cell-targeting Nr5a1-Cre mice were crossed with Tspo-floxed mice. The resulting Tspo(fl/fl);Nr5a1-Cre(/+) mice were born at a normal Mendelian ratio. Nr5a1-driven Tspo cKO mice exhibited highly reduced Tspo levels in adrenal cortex and gonads. Treatment of mice with human chorionic gonadotropin (hCG) resulted in increased circulating testosterone levels despite extensive lipid droplet depletion. In contrast, Nr5a1-driven Tspo cKO mice lost their ability to form corticosterone in response to adrenocorticotropic hormone (ACTH). Important for ACTH-dependent steroidogenesis, Mc2r, Stard1, and Cypa11a1 levels were unaffected, whereas Scarb1 levels were increased and accumulation of lipid droplets was observed, indicative of a blockade of cholesterol utilization for steroidogenesis. TSPO expression in the adrenal medulla and increased epinephrine production were also observed. In conclusion, TSPO was found necessary for preimplantation embryo development and ACTH-stimulated steroid biosynthesis.


Subject(s)
Corticosterone/biosynthesis , Receptors, GABA/physiology , Testosterone/biosynthesis , Animals , Brain/metabolism , Female , Gene Expression Regulation , Gene Silencing , Gonads/metabolism , Male , Mice , Mice, Knockout , Receptors, GABA/genetics , Receptors, GABA/metabolism , Stress, Physiological
6.
Pharmacol Res ; 99: 393-403, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25936508

ABSTRACT

Mitochondria play a critical role in the physiological homeostasis of the cell, contributing to numerous cellular processes, including bioenergetics, metabolism and cell life and death. Owing to their keystone role, mitochondria have gained much attention as pharmacological targets. The outer mitochondrial integral membrane translocator protein (TSPO) has attracted a significant degree of pharmacological interest owing to its ability to bind a number of classes of drugs with high affinity and specificity. In addition to its well-characterized drug binding site, TSPO possess an additional high-affinity ligand binding site, originally identified for its ability to bind the lipid cholesterol, which was named the cholesterol recognition/interaction amino acid consensus (CRAC) motif. Previous investigations from our laboratory identified additional ligands targeted to TSPO's CRAC motif which are able to potently inhibit mitochondrial cholesterol transport and steroid biosynthesis, processes for which TSPO has been well-characterized. However, all of these compounds possessed the steroidal backbone common to cholesterol and steroid hormones. In our efforts to expand our understanding of TSPO's CRAC motif, we performed studies aimed at identifying non-steroidal ligands for this motif. Molecular modeling and in silico screening of large chemical libraries identified a panel of compounds which were subsequently screened for bioactivity in a number of steroidogenic model systems. These efforts identified a family of non-steroidal CRAC ligands able to potently inhibit steroidogenesis, and at higher concentrations, promote apoptosis. In addition, the best candidate in this family was able to suppress testosterone synthesis when administered to rats, indicating that this novel family of non-steroidal CRAC ligands may serve as prototypes for the development of drugs useful for treatment of diseases of steroid overproduction, such as Cushing's syndrome and steroidogenic cell tumors in humans and animals.


Subject(s)
Amino Acids/metabolism , Cholesterol/metabolism , Steroids/metabolism , Animals , Binding Sites , Biological Transport/physiology , Consensus , Ligands , Mice , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/metabolism , Models, Molecular , Protein Structure, Tertiary , Protein Transport/physiology , Rats
7.
Eur J Pharm Sci ; 76: 231-7, 2015 Aug 30.
Article in English | MEDLINE | ID: mdl-26002041

ABSTRACT

Ligands of 18-kDa translocator protein (TSPO) are known for their ability to potently and dose-dependently stimulate steroid biosynthesis in steroidogenic cells. In this study, we investigated a number of 2-phenyl-imidazo[1,2-a]pyridine acetamide derivatives, analogs of alpidem, for their ability to bind TSPO and to affect steroidogenesis in a mouse Leydig tumor cell line. We observed that not only some compounds behaved as agonists, stimulating steroidogenesis (e.g., 3 and 4) with EC50 values (15.9 and 6.99µM) close to that determined for FGIN-1-27 used as positive control (7.24µM), but two compounds, namely 5 and 6, which on the other hand are the most lipophilic ones in the investigated series, behaved as antagonists, by significantly inhibiting steroid production at concentrations at least twenty times lower than the cytotoxic ones. To our surprise, the newly synthesized compound 3, which is a strict analog of alpidem bearing at the para position of the 2-phenyl group a methoxy group instead of chlorine, achieved a ten-fold stimulation of the steroid production (for comparison FGIN-1-27 achieved 1.6-fold stimulation). Within the limits of the examined property space, some unprecedented SARs were unveiled, which can help in understanding the key molecular factors underlying the transition from agonism to antagonism in the steroidogenesis process. Besides the substitution pattern and the physicochemical features (mainly hydrogen bonding potential) of the substituents at the positions C(6) and C(8) of the imidazo[1,2-a]pyridine nucleus, and at the para position of the 2-phenyl group, the structure-activity relationship analysis suggested lipophilicity, whose increase seems to be generally related to steroidogenesis inhibition, and steric hindrance, which appeared as a stimulation-limiting factor, as two main properties to control in the design or optimization of novel imidazo[1,2-a]pyridine-based TSPO ligands endowed with potential in modulating the steroidogenesis process.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Hormone Antagonists/pharmacology , Imidazoles/pharmacology , Leydig Cell Tumor/drug therapy , Neoplasms, Hormone-Dependent/drug therapy , Pyridines/pharmacology , Receptors, GABA/drug effects , Steroid Synthesis Inhibitors/pharmacology , Testicular Neoplasms/drug therapy , Animals , Antineoplastic Agents, Hormonal/chemical synthesis , Cell Line, Tumor , Dose-Response Relationship, Drug , Hormone Antagonists/chemical synthesis , Hydrogen Bonding , Imidazoles/chemical synthesis , Leydig Cell Tumor/metabolism , Leydig Cell Tumor/pathology , Ligands , Male , Mice , Molecular Structure , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Pyridines/chemical synthesis , Receptors, GABA/metabolism , Steroid Synthesis Inhibitors/chemical synthesis , Structure-Activity Relationship , Testicular Neoplasms/metabolism , Testicular Neoplasms/pathology
8.
Mol Cell Endocrinol ; 408: 90-8, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25818881

ABSTRACT

Steroidogenesis begins with cholesterol transfer into mitochondria through the transduceosome, a complex composed of cytosolic proteins that include steroidogenesis acute regulatory protein (STAR), 14-3-3 adaptor proteins, and the outer mitochondrial membrane proteins Translocator Protein (TSPO) and Voltage-Dependent Anion Channel (VDAC). TSPO is a drug- and cholesterol-binding protein found at particularly high levels in steroid synthesizing cells. Its aberrant expression has been linked to cancer, neurodegeneration, neuropsychiatric disorders and primary hypogonadism. Brain steroids serve as local regulators of neural development and excitability. Reduced levels of these steroids have been linked to depression, anxiety and neurodegeneration. Reduced serum testosterone is common among subfertile young men and aging men, and is associated with depression, metabolic syndrome and reduced sexual function. Although testosterone-replacement therapy is available, there are undesired side-effects. TSPO drug ligands have been proposed as therapeutic agents to regulate steroid levels in the brain and testis.


Subject(s)
Cholesterol/metabolism , Receptors, GABA/metabolism , Steroids/biosynthesis , Animals , Biological Transport , Humans , Models, Biological , Molecular Targeted Therapy
9.
Biol Reprod ; 91(4): 96, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25210128

ABSTRACT

Mitochondria are home to many cellular processes, including oxidative phosphorylation and fatty acid metabolism, and in steroid-synthesizing cells, they are involved in cholesterol import and metabolism, which is the initiating step in steroidogenesis. The formation of macromolecular protein complexes aids in the regulation and efficiency of these mitochondrial functions, though because of their dynamic nature, they are hard to identify. To overcome this problem, we used Blue-Native PAGE with whole-gel mass spectrometry on isolated mitochondria from control and hormone-treated MA-10 mouse tumor Leydig cells. The presence of multiple mitochondrial protein complexes was shown. Although these were qualitatively similar under control and human chorionic gonadotropin (hCG)-stimulated conditions, quantitative differences in the components of the complexes emerged after hCG treatment. A prominent decrease was observed with proteins involved in fatty acid import into the mitochondria, implying that mitochondrial beta-oxidation is not essential for steroidogenesis. To confirm this observation, we inhibited fatty acid import utilizing the CPT1a inhibitor etomoxir, resulting in increased steroid production. Conversely, stimulation of mitochondrial beta-oxidation with metformin resulted in a dose-dependent reduction in steroidogenesis. These changes were accompanied by changes in mitochondrial respiration and in the lactic acid formed during glycolysis. Taken together, these results suggest that upon hormonal stimulation, mitochondria efficiently import cholesterol for steroid production at the expense of other lipids necessary for energy production, specifically fatty acids required for beta-oxidation.


Subject(s)
Fatty Acids/metabolism , Leydig Cells/metabolism , Mitochondria/metabolism , Steroids/biosynthesis , Animals , Biological Transport , Cell Line , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Hypoglycemic Agents/pharmacology , Male , Metformin/pharmacology , Mice , Oxidation-Reduction , Transcriptome
10.
Traffic ; 15(9): 895-914, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24890942

ABSTRACT

Steroid hormones, bioactive oxysterols and bile acids are all derived from the biological metabolism of lipid cholesterol. The enzymatic pathways generating these compounds have been an area of intense research for almost a century, as cholesterol and its metabolites have substantial impacts on human health. Owing to its high degree of hydrophobicity and the chemical properties that it confers to biological membranes, the distribution of cholesterol in cells is tightly controlled, with subcellular organelles exhibiting highly divergent levels of cholesterol. The manners in which cells maintain such sterol distributions are of great interest in the study of steroid and bile acid synthesis, as limiting cholesterol substrate to the enzymatic pathways is the principal mechanism by which production of steroids and bile acids is regulated. The mechanisms by which cholesterol moves within cells, however, remain poorly understood. In this review, we examine the subcellular machinery involved in cholesterol metabolism to steroid hormones and bile acid, relating it to both lipid- and protein-based mechanisms facilitating intracellular and intraorganellar cholesterol movement and delivery to these pathways. In particular, we examine evidence for the involvement of specific protein domains involved in cholesterol binding, which impact cholesterol movement and metabolism in steroidogenesis and bile acid synthesis. A better understanding of the physical mechanisms by which these protein- and lipid-based systems function is of fundamental importance to understanding physiological homeostasis and its perturbation.


Subject(s)
Bile Acids and Salts/metabolism , Biological Transport/physiology , Gonadal Steroid Hormones/metabolism , Protein Structure, Tertiary/physiology , Sterols/metabolism , Animals , Carrier Proteins/metabolism , Cholesterol/metabolism , Homeostasis/physiology , Humans , Lipid Metabolism/physiology
11.
J Med Chem ; 56(12): 5115-29, 2013 Jun 27.
Article in English | MEDLINE | ID: mdl-23725591

ABSTRACT

Inhibition of GSK-3ß has been well documented to account for the behavioral actions of the mood stabilizer lithium in various animal models of mood disorders. Recent studies have showed that genetic or pharmacological inhibition of GSK-3ß resulted in anxiolytic-like and pro-social behavior. In our ongoing efforts to develop GSK-3ß inhibitors for the treatment of mood disorders, SAR studies on maleimide-based compounds were undertaken. We present herein for the first time that some of these GSK-3ß inhibitors, in particular analogues 1 and 9, were able to stimulate progesterone production in the MA-10 mouse tumor Leydig cell model of steroidogenesis without any significant toxicity. These two compounds were tested in the SmartCube behavioral assay and showed anxiolytic-like signatures following daily dose administration (50 mg/kg, ip) for 13 days. Taken together, these results support the hypothesis that GSK-3ß inhibition could influence neuroactive steroid production thereby mediating the modulation of anxiety-like behavior in vivo.


Subject(s)
Glycogen Synthase Kinase 3/antagonists & inhibitors , Maleimides/chemistry , Maleimides/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Steroids/biosynthesis , Adenosine Triphosphate/metabolism , Animals , Binding, Competitive , Cell Line, Tumor , Humans , Maleimides/metabolism , Mice , Protein Kinase Inhibitors/metabolism
12.
Steroids ; 77(13): 1327-34, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22986075

ABSTRACT

Steroids metabolically derive from lipid cholesterol, and vertebrate steroids additionally derive from the steroid pregnenolone. Pregnenolone is derived from cholesterol by hydrolytic cleavage of the aliphatic tail by mitochondrial cytochrome P450 enzyme CYP11A1, located in the inner mitochondrial membrane. Delivery of cholesterol to CYP11A1 comprises the principal control step of steroidogenesis, and requires a series of proteins spanning the mitochondrial double membranes. A critical member of this cholesterol translocation machinery is the integral outer mitochondrial membrane translocator protein (18kDa, TSPO), a high-affinity drug- and cholesterol-binding protein. The cholesterol-binding site of TSPO consists of a phylogenetically conserved cholesterol recognition/interaction amino acid consensus (CRAC). Previous studies from our group identified 5-androsten-3ß,17,19-triol (19-Atriol) as drug ligand for the TSPO CRAC motif inhibiting cholesterol binding to CRAC domain and steroidogenesis. To further understand 19-Atriol's mechanism of action as well as the molecular recognition by the TSPO CRAC motif, we undertook structure-activity relationship (SAR) analysis of the 19-Atriol molecule with a variety of substituted steroids oxygenated at positions around the steroid backbone. We found that in addition to steroids hydroxylated at carbon C19, hydroxylations at C4, C7, and C11 contributed to inhibition of cAMP-mediated steroidogenesis in a minimal steroidogenic cell model. However, only substituted steroids with C19 hydroxylations exhibited specificity to TSPO, its CRAC motif, and mitochondrial cholesterol transport, as the C4, C7, and C11 hydroxylated steroids inhibited the metabolic transformation of cholesterol by CYP11A1. We thus provide new insights into structure-activity relationships of steroids inhibiting mitochondrial cholesterol transport and steroidogenic cholesterol metabolic enzymes.


Subject(s)
Steroids/biosynthesis , Steroids/pharmacology , Animals , Cell Line, Tumor , Cell Survival/drug effects , Hydroxylation , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Oxygen/chemistry , Receptors, GABA/metabolism , Steroids/chemistry , Steroids/metabolism , Structure-Activity Relationship
13.
Mol Endocrinol ; 26(11): 1868-82, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22973050

ABSTRACT

Steroid hormones are critical for organismal development and health. The rate-limiting step in steroidogenesis is the transport of cholesterol from the outer mitochondrial membrane (OMM) to the cytochrome P450 enzyme CYP11A1 in the inner mitochondrial membrane (IMM). Cholesterol transfer occurs through a complex termed the "transduceosome," in which cytosolic steroidogenic acute regulatory protein interacts with OMM proteins translocator protein and voltage-dependent anion channel (VDAC) to assist with the transfer of cholesterol to OMM. It has been proposed that cholesterol transfer from OMM to IMM occurs at specialized contact sites bridging the two membranes composed of VDAC and IMM adenine nucleotide translocase (ANT). Blue native PAGE of Leydig cell mitochondria identified two protein complexes that were able to bind cholesterol at 66- and 800-kDa. Immunoblot and mass spectrometry analyses revealed that the 800-kDa complex contained the OMM translocator protein (18-kDa) and VDAC along with IMM CYP11A1, ATPase family AAA domain-containing protein 3A (ATAD3A), and optic atrophy type 1 proteins, but not ANT. Knockdown of ATAD3A, but not ANT or optic atrophy type 1, in Leydig cells resulted in a significant decrease in hormone-induced, but not 22R-hydroxycholesterol-supported, steroid production. Using a 22-phenoxazonoxy-5-cholene-3-beta-ol CYP11A1-specific probe, we further demonstrated that the 800-kDa complex offers the microenvironment needed for CYP11A1 activity. Addition of steroidogenic acute regulatory protein to the complex mobilized the cholesterol bound at the 800-kDa complex, leading to increased steroid formation. These results identify a bioactive, multimeric protein complex spanning the OMM and IMM unit that is responsible for the hormone-induced import, segregation, targeting, and metabolism of cholesterol.


Subject(s)
Cholesterol/metabolism , Hormones/metabolism , Mitochondrial Proteins/metabolism , Multiprotein Complexes/metabolism , Animals , Biological Transport/drug effects , Cholesterol/chemistry , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Chorionic Gonadotropin/pharmacology , Hormones/pharmacology , Leydig Cells/metabolism , Leydig Cells/ultrastructure , Male , Mass Spectrometry , Mice , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/metabolism , Models, Biological , Molecular Weight , Native Polyacrylamide Gel Electrophoresis , Oxazines/chemistry , Oxazines/metabolism , Phosphoproteins/metabolism
14.
Bioorg Med Chem Lett ; 22(12): 4139-43, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22575868

ABSTRACT

A homology model of the steroidogenic acute regulatory protein (STAR)-related lipid transfer (START) domain of STARD1 was built, and the cholesterol binding site was identified. Structure-based design studies were performed to identify small molecule inhibitors of the START domain. The lead compounds were selected based on cAMP-induced, but not 22R-hydroxycholesterol-supported, inhibition of steroid synthesis by 50% at 10 µM. The results obtained by molecular docking & dynamics show a good correlation between bioactivity, docking scores and calculated binding energies of ligand-protein complexes. The best active compounds will be optimized further and used to develop potential drugs to control excessive steroid formation.


Subject(s)
Cholesterol/chemistry , Cyclic AMP/chemistry , Phosphoproteins/antagonists & inhibitors , Small Molecule Libraries/chemical synthesis , Amino Acid Sequence , Animals , Binding Sites , Cholesterol/metabolism , Drug Design , Humans , Kinetics , Leydig Cells/drug effects , Leydig Cells/metabolism , Ligands , Male , Mice , Molecular Dynamics Simulation , Molecular Sequence Data , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Protein Binding , Protein Structure, Tertiary , Small Molecule Libraries/pharmacology , Structural Homology, Protein , Structure-Activity Relationship , Thermodynamics
15.
J Biol Chem ; 286(11): 9875-87, 2011 Mar 18.
Article in English | MEDLINE | ID: mdl-21209087

ABSTRACT

Steroid hormones are metabolically derived from multiple enzymatic transformations of cholesterol. The controlling step in steroid hormone biogenesis is the delivery of cholesterol from intracellular stores to the cytochrome P450 enzyme CYP11A1 in the mitochondrial matrix. The 18-kDa translocator protein (TSPO) plays an integral part in this mitochondrial cholesterol transport. Consistent with its role in intracellular cholesterol movement, TSPO possesses a cholesterol recognition/interaction amino acid consensus (CRAC) motif that has been demonstrated to bind cholesterol. To further investigate the TSPO CRAC motif, we performed molecular modeling studies and identified a novel ligand, 3,17,19-androsten-5-triol (19-Atriol) that inhibits cholesterol binding at the CRAC motif. 19-Atriol could bind a synthetic CRAC peptide and rapidly inhibited hormonally induced steroidogenesis in MA-10 mouse Leydig tumor cells and constitutive steroidogenesis in R2C rat Leydig tumor cells at low micromolar concentrations. Inhibition at these concentrations was not due to toxicity or inhibition of the CYP11A1 enzyme and was reversed upon removal of the compound. In addition, 19-Atriol was an even more potent inhibitor of PK 11195-stimulated steroidogenesis, with activity in the high nanomolar range. This was accomplished without affecting PK 11195 binding or basal steroidogenesis. Finally, 19-Atriol inhibited mitochondrial import and processing of the steroidogenic acute regulatory protein without any effect on TSPO protein levels. In conclusion, we have identified a novel androstenetriol that can interact with the CRAC domain of TSPO, can control hormonal and constitutive steroidogenesis, and may prove to be a useful tool in the therapeutic control of diseases of excessive steroid formation.


Subject(s)
Androstenols/chemistry , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Cholesterol/metabolism , Mitochondria/metabolism , Peptides/chemistry , Peptides/metabolism , Receptors, GABA-A/chemistry , Receptors, GABA-A/metabolism , Receptors, GABA/metabolism , Amino Acid Motifs , Androstenols/pharmacology , Animals , Carrier Proteins/genetics , Cell Line, Tumor , Cholesterol Side-Chain Cleavage Enzyme/genetics , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Dose-Response Relationship, Drug , Mice , Mitochondria/chemistry , Mitochondria/genetics , Peptides/genetics , Protein Binding/drug effects , Protein Structure, Tertiary , Rats , Receptors, GABA/chemistry , Receptors, GABA/genetics , Receptors, GABA-A/genetics
16.
Biol Reprod ; 84(5): 976-85, 2011 May.
Article in English | MEDLINE | ID: mdl-21228212

ABSTRACT

Previous studies in MA-10 tumor Leydig cells demonstrated that disruption of the mitochondrial electron-transport chain (ETC), membrane potential (ΔΨ(m)), or ATP synthesis independently inhibited steroidogenesis. In contrast, studies of primary Leydig cells indicated that the ETC, ΔΨ(m), and ATP synthesis cooperatively affected steroidogenesis. These results suggest significant differences between the two systems and call into question the extent to which results from tumor Leydig cells relate to primary cells. Thus, to further understand the similarities and differences between the two systems as well as the impact of ATP disruption on steroidogenesis, we performed comparative studies of MA-10 and primary Leydig cells under similar conditions of mitochondrial disruption. We show that mitochondrial ATP synthesis is critical for steroidogenesis in both primary and tumor Leydig cells. However, in striking contrast to primary cells, perturbation of ΔΨ(m) in MA-10 cells did not substantially decrease cellular ATP content, a perplexing finding because ΔΨ(m) powers the mitochondrial ATP synthase. Further studies revealed that a significant proportion of cellular ATP in MA-10 cells derives from glycolysis. In contrast, primary cells appear to be almost completely dependent on mitochondrial respiration for their energy provision. Inhibitor studies also suggested that the MA-10 ETC is impaired. This work underscores the importance of mitochondrial ATP for hormone-stimulated steroid production in both MA-10 and primary Leydig cells while indicating that caution must be exercised in extrapolating data from tumor cells to primary tissue.


Subject(s)
Adenosine Triphosphate/metabolism , Cell Line, Tumor , Cells, Cultured , Leydig Cell Tumor/metabolism , Leydig Cells/metabolism , Mitochondria/metabolism , Steroids/biosynthesis , Animals , Electron Transport Chain Complex Proteins/antagonists & inhibitors , Electron Transport Chain Complex Proteins/metabolism , Enzyme Inhibitors/pharmacology , Glycolysis/drug effects , Kinetics , Laser Scanning Cytometry , Leydig Cells/cytology , Leydig Cells/pathology , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/drug effects , Progesterone/metabolism , Rats , Testicular Neoplasms/metabolism , Testosterone/metabolism , Uncoupling Agents/pharmacology
17.
Mol Cell Endocrinol ; 336(1-2): 70-9, 2011 Apr 10.
Article in English | MEDLINE | ID: mdl-21147195

ABSTRACT

The principal site of regulation of steroid hormone biosynthesis is the transfer of cholesterol from the outer to inner mitochondrial membrane. Hormonal stimulation of steroidogenic cells promotes this mitochondrial lipid import through a multi-protein complex, termed the transduceosome, spanning the two membranes. The transduceosome complex is assembled from multiple proteins, such as the steroidogenic acute regulatory (STAR) protein and translocator protein (TSPO), and requires their targeting to the mitochondria for transduceosome function. The vast majority of mitochondrial proteins, including those participating in cholesterol import, are encoded in the nucleus. Their subsequent mitochondrial incorporation is performed through a series of protein import machineries located in the outer and inner mitochondrial membranes. Here we review our current knowledge of the mitochondrial cholesterol import machinery of the transduceosome. This is complemented with descriptions of mitochondrial protein import machineries and mechanisms by which these machineries assemble the transduceosome in steroidogenic mitochondria.


Subject(s)
Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Steroids/metabolism , Animals , Humans , Protein Processing, Post-Translational , Protein Transport
18.
Cancer Biol Ther ; 8(1): 94-100, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19127118

ABSTRACT

MED1 is a base excision repair enzyme that interacts with the mismatch repair protein MLH1 and maintains genomic integrity by binding methylated DNA and repairing spontaneous deamination events. MED1 mutations have been associated with microsatellite instability and accelerated colorectal cancer (CRC) tumorigenesis. We propose that promoter methylation may serve as an alternative epigenetic mechanism for MED1 gene suppression during sporadic CRC tumorigenesis. Methylation status of the MED1 promoter was investigated in a panel of ovarian and colorectal cancer cell lines. The MED1 promoter region was sequenced following bisulfite treatment and sequence analysis identified a CpG island within the MED1 promoter which is frequently and preferentially methylated (> or =50%) in ovarian and colorectal cancer cell lines with low/reduced MED1 expression. In vitro reversal of methylation restored MED1 expression. In colorectal cancer patients, when MED1 methylation was present, both tumor and matched mucosa were affected equally (mean frequency of methylation 24%) and there was no correlation between methylation and tumor stage. Patients without history of CRC showed significantly lower frequency of methylation (mean 14%, p < 0.05). Decreased MED1 transcript levels were observed in matched normal mucosa when compared to controls (median fold difference 8.0). Additional decreased expression was seen between mucosa and matched tumor (median fold decrease 4.4). Thus, MED1 promoter methylation and gene silencing occur in sporadic CRC patients and represent an early event in CRC tumorigenesis. Detection of MED1 methylation and gene suppression in normal colon mucosa may contribute to identifying patients at higher risk of developing CRC during screening procedures.


Subject(s)
Colorectal Neoplasms/genetics , DNA Repair/genetics , Endodeoxyribonucleases/genetics , Ovarian Neoplasms/genetics , Transcription Factors/genetics , Base Pair Mismatch , DNA Methylation , DNA, Neoplasm/genetics , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Humans , Mediator Complex Subunit 1
19.
Mol Cell Endocrinol ; 299(1): 23-31, 2009 Feb 05.
Article in English | MEDLINE | ID: mdl-18761053

ABSTRACT

In males, serum testosterone levels decline with advancing age. Though part of a complex process, this age-related decline in testosterone appears to occur, in part, due to a significant decline in the ability of aged Leydig cells to produce testosterone maximally in response to luteinizing hormone (LH). The structure of the molecular machinery responsible for the synthesis of testosterone is described, and placed in the context of Leydig cell biology. Multiple parameters related to the synthesis of testosterone by the Leydig cell have been observed to change with age. Relationships among these changes are reviewed. A discussion of potential causes of the age-related decline in Leydig cell steroidogenic capacity presents a model in which the inability of aged cells to adequately respond to hormonal stimulation results in cellular regression with concomitant decline in maximal testosterone output.


Subject(s)
Cellular Senescence/physiology , Leydig Cells/physiology , Testosterone/biosynthesis , Animals , Down-Regulation/physiology , Humans , Leydig Cells/metabolism , Male , Models, Biological , Signal Transduction/physiology
20.
Endocrinology ; 148(6): 2583-90, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17332065

ABSTRACT

Studies of MA-10 Leydig cells have shown that intact mitochondria with active respiration are essential for LH-induced Leydig cell steroidogenesis. To further elucidate the role played by mitochondria in steroidogenesis, we examined the effects of the perturbation of the mitochondrial electron transport chain with myxothiazol (MYX) on testosterone production by primary cultures of Brown Norway rat Leydig cells. Analysis of the steroidogenic pathway revealed that cAMP production and the activities of each of 3beta-hydroxysteroid dehydrogenase, 17alpha-hydroxylase/C17-20 lyase, and 17beta-hydroxysteroid dehydrogenase were inhibited by MYX and that LH-stimulated testosterone production was suppressed. In contrast to the inhibition of LH-stimulated testosterone production by MYX, the incubation of Leydig cells with MYX in the absence of LH stimulated testosterone production. Although testosterone production was increased, steroidogenic acute regulatory protein was decreased in response to MYX, not increased as could be expected. Additional electron transport chain inhibitors had stimulatory effects on testosterone production that were similar to those of MYX, strongly suggesting that the effect of MYX on basal testosterone production is related to its effect on the mitochondrial electron transport chain. Finally, incubation of the cells with a combination of MYX and the calcium chelator 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetracetic acid tetrakis acetoxymethyl ester suppressed MYX-mediated increased basal steroidogenesis but had no effect on hydroxycholesterol-mediated steroidogenesis. Taken together, these results indicate that inhibition of the mitochondrial electron transport chain can block LH-stimulated testosterone production through suppression of a number of steps of the steroidogenic pathway but also stimulates basal testosterone production through a calcium-mediated mechanism.


Subject(s)
Leydig Cells/drug effects , Leydig Cells/metabolism , Luteinizing Hormone/pharmacology , Steroids/biosynthesis , Testosterone/biosynthesis , Animals , Male , Methacrylates/pharmacology , Mitochondria/drug effects , Mitochondria/physiology , Rats , Rats, Inbred Strains , Signal Transduction , Thiazoles/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL